Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 10(1): 5647, 2020 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-32221352

RESUMEN

During mammalian development, establishing functional neural networks in stratified tissues of the mammalian central nervous system depends upon the proper migration and positioning of neurons, a process known as lamination. In particular, the pseudostratified neuroepithelia of the retina and cerebrocortical ventricular zones provide a platform for progenitor cell proliferation and migration. Lamination defects in these tissues lead to mispositioned neurons, disrupted neuronal connections, and abnormal function. The molecular mechanisms necessary for proper lamination in these tissues are incompletely understood. Here, we identified a nonsense mutation in the Eml1 gene in a novel murine model, tvrm360, displaying subcortical heterotopia, hydrocephalus and disorganization of retinal architecture. In the retina, Eml1 disruption caused abnormal positioning of photoreceptor cell nuclei early in development. Upon maturation, these ectopic photoreceptors possessed cilia and formed synapses but failed to produce robust outer segments, implying a late defect in photoreceptor differentiation secondary to mislocalization. In addition, abnormal positioning of Müller cell bodies and bipolar cells was evident throughout the inner neuroblastic layer. Basal displacement of mitotic nuclei in the retinal neuroepithelium was observed in tvrm360 mice at postnatal day 0. The abnormal positioning of retinal progenitor cells at birth and ectopic presence of photoreceptors and secondary neurons upon maturation suggest that EML1 functions early in eye development and is crucial for proper retinal lamination during cellular proliferation and development.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/fisiología , Proteínas Asociadas a Microtúbulos/metabolismo , Retina/metabolismo , Retina/fisiología , Animales , Proliferación Celular/fisiología , Células Ependimogliales/metabolismo , Ratones , Ratones Endogámicos C57BL , Neurogénesis/fisiología , Células Fotorreceptoras Retinianas Conos/metabolismo , Células Madre/metabolismo , Células Madre/fisiología
2.
Hum Mol Genet ; 14(16): 2323-33, 2005 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-16000322

RESUMEN

Mutations in the human ALMS1 gene cause Alström syndrome (AS), a progressive disease characterized by neurosensory deficits and by metabolic defects including childhood obesity, hyperinsulinemia and Type 2 diabetes. Other features that are more variable in expressivity include dilated cardiomyopathy, hypertriglyceridemia, hypercholesterolemia, scoliosis, developmental delay and pulmonary and urological dysfunctions. ALMS1 encodes a ubiquitously expressed protein of unknown function. To obtain an animal model in which the etiology of the observed pathologies could be further studied, we generated a mouse model using an Alms1 gene-trapped ES cell line. Alms1-/- mice develop features similar to patients with AS, including obesity, hypogonadism, hyperinsulinemia, retinal dysfunction and late-onset hearing loss. Insulin resistance and increased body weight are apparent between 8 and 12 weeks of age, with hyperglycemia manifesting at approximately 16 weeks of age. In addition, Alms1-/- mice have normal hearing until 8 months of age, after which they display abnormal auditory brainstem responses. Diminished cone ERG b-wave response is observed early, followed by the degeneration of photoreceptor cells. Electron microscopy revealed accumulation of intracellular vesicles in the inner segments of photoreceptors, whereas immunohistochemical analysis showed mislocalization of rhodopsin to the outer nuclear layer. These findings suggest that ALMS1 has a role in intracellular trafficking.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Modelos Animales de Enfermedad , Degeneración Nerviosa/genética , Obesidad/genética , Proteínas/fisiología , Degeneración Retiniana/genética , Animales , Proteínas de Ciclo Celular , Electrorretinografía , Femenino , Pérdida Auditiva/genética , Humanos , Hiperinsulinismo/genética , Resistencia a la Insulina/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transporte de Proteínas/genética , Proteínas/genética , Homología de Secuencia de Aminoácido , Síndrome
3.
Hum Mol Genet ; 10(16): 1619-26, 2001 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-11487564

RESUMEN

The rd7 mouse is a model for hereditary retinal degeneration characterized clinically by retinal spotting throughout the fundus and late onset retinal degeneration, and histologically by retinal dysplasia manifesting as folds and whorls in the photoreceptor layer. This study demonstrates that the rd7 phenotype results from a splicing error created by a genomic deletion of an intron and part of an exon. Hematoxylin/eosin staining of rd7 tissue shows that the whorls in the outer nuclear layer of the retina do not appear during embryonic development but manifest by postnatal day 12.5 (P12.5). Furthermore, in situ hybridization data indicates that the Nr2e3 message is first present at barely discernable levels at embryonic day 18.5, becomes abundant by P2.5, and reaches maximal adult levels by P10.5. Results from these experiments indicate that Nr2e3 message is expressed prior to the development of S-cones. This data coincides with studies in humans showing that mutations in Nr2e3 result in a unique type of retinal degeneration known as enhanced S-cone syndrome, where patients have a 30-fold increase in S-cone sensitivity compared to normal. Immunohistochemical staining of cone cells demonstrates that rd7 retinas have an increased number of cone cells compared to wild-type retinas. Thus, Nr2e3 may function by regulating genes involved in cone cell proliferation, and mutations in this gene lead to retinal dysplasia and degeneration by disrupting normal photoreceptor cell topography as well as cell-cell interactions.


Asunto(s)
Receptores Citoplasmáticos y Nucleares/fisiología , Células Fotorreceptoras Retinianas Conos/citología , Degeneración Retiniana/etiología , Displasia Retiniana/etiología , Factores de Transcripción/fisiología , Animales , Recuento de Células , Diferenciación Celular/genética , Diferenciación Celular/fisiología , División Celular/genética , División Celular/fisiología , Femenino , Fondo de Ojo , Perfilación de la Expresión Génica , Genotipo , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Mutación , Receptores Nucleares Huérfanos , Fenotipo , Receptores Citoplasmáticos y Nucleares/genética , Retina/metabolismo , Degeneración Retiniana/genética , Displasia Retiniana/genética , Factores de Transcripción/genética
4.
Hum Mol Genet ; 10(12): 1325-34, 2001 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-11406614

RESUMEN

The tubby-like protein 3 (Tulp3) gene has been identified as a member of a small novel gene family which is primarily neuronally expressed. Mutations in two of the family members, tub and tulp1, have been shown to cause neurosensory disorders. To determine the in vivo function of Tulp3, we have generated a germline mutation in the mouse Tulp3 gene by homologous recombination. Embryos homozygous for the Tulp3 mutant allele exhibit failure of neural tube closure, and die by embryonic day 14.5. Failure of cranial neural tube closure coincided with increased neuroepithelial apoptosis specifically in the hindbrain and the caudal neural tube. In addition, the number of betaIII-tubulin positive cells is significantly decreased in the hindbrain of Tulp3(-/-) embryos. These results suggest that disruption of the Tulp3 gene affects the development of a neuronal cell population. Interestingly, some Tulp3 heterozygotes also manifest embryonic lethality with neuroepithelial cell death. Our results demonstrate that the Tulp3 gene is essential for embryonic development in mice.


Asunto(s)
Defectos del Tubo Neural/genética , Proteínas/genética , Animales , Apoptosis , Diferenciación Celular , Desarrollo Embrionario y Fetal , Células Epiteliales/patología , Marcadores Genéticos , Heterocigoto , Péptidos y Proteínas de Señalización Intercelular , Péptidos y Proteínas de Señalización Intracelular , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Mutación , Defectos del Tubo Neural/embriología , Defectos del Tubo Neural/patología , Neuronas/metabolismo , Fenotipo
5.
Genomics ; 74(2): 219-27, 2001 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-11386758

RESUMEN

The murine Lbx2 gene is a member of the ladybird family of homeobox genes, which is expressed in the developing urogenital system, eye, and brain. Using transgenic mice, we demonstrate that 9 kb of the 5' flanking region of mouse Lbx2 is able to direct expression of a reporter gene in a tissue-specific manner recapitulating the endogenous expression pattern. This regulatory region provides a novel reagent allowing for transgenic expression in the developing urogenital ridge. In addition, we describe the identification of the human homologue, LBX2. Comparison of the human LBX2 and mouse Lbx2 sequences upstream of the coding regions reveals sequence conservation suggesting conserved regulatory regions. Both the human LBX2 and the mouse Lbx2 genes have similar genomic structures and are composed of two exons separated by an intron. We mapped the mouse Lbx2 gene to 35 cM on chromosome 6 and the human LBX2 gene to a homologous region of chromosome 2p13. This is a candidate region for several inherited disorders, including Alström syndrome, a disorder that includes ocular, urogenital, and renal abnormalities. Given the expression pattern of Lbx2, the chromosomal location in humans, and the potential function of mammalian ladybird genes, we have begun to analyze patients with ocular disorders and those with Alström syndrome for mutations in LBX2. Although polymorphisms were identified, our results indicate that mutations in the coding region of LBX2 do not account for Alström syndrome in the six kindreds analyzed.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Pérdida Auditiva Sensorineural/genética , Proteínas de Homeodominio/genética , Hipogonadismo/genética , Enfermedades Renales/genética , Mutación , Obesidad/genética , Regiones Promotoras Genéticas , Enfermedades de la Retina/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Mapeo Cromosómico , Clonación Molecular , Análisis Mutacional de ADN , Ojo/embriología , Oftalmopatías/genética , Salud de la Familia , Femenino , Genes Recesivos , Genes Reporteros , Pruebas Genéticas , Humanos , Hibridación in Situ , Intrones , Masculino , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Linaje , Homología de Secuencia de Aminoácido , Homología de Secuencia de Ácido Nucleico , Síndrome , Distribución Tisular
6.
Genomics ; 74(3): 273-86, 2001 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-11414755

RESUMEN

The TallyHo (TH) mouse strain is a newly established model for non-insulin-dependent diabetes mellitus (NIDDM). TH mice show obesity, hyperinsulinemia, hyperlipidemia, and male-limited hyperglycemia. A genetic dissection of the diabetes syndrome has been carried out using male backcross 1 progeny obtained from crosses between (C57BL/6J x TH)F1 and TH mice or (CAST/Ei x TH)F1 and TH mice. A genome-wide scan reveals three quantitative trait loci (QTLs), Tanidd1-3 (TH-associated NIDDM) linked to hyperglycemia. The major QTL (common in both crosses), Tanidd1, maps to chromosome (Chr) 19. Additionally, gene-gene interactions contributing to hyperglycemia have been observed between Tanidd1 and a locus on Chr 18 as well as between Tanidd2 and a locus on Chr 16. The overt hyperglycemia in TH mice is, therefore, likely due to a mutation in a major diabetes susceptibility locus on Chr 19, which interacts with additional genes to lead to an observable phenotype.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Modelos Animales de Enfermedad , Animales , Mapeo Cromosómico , Cruzamientos Genéticos , Diabetes Mellitus Tipo 2/patología , Femenino , Predisposición Genética a la Enfermedad/genética , Genoma , Hiperglucemia/genética , Hiperlipidemias/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Obesidad/genética , Carácter Cuantitativo Heredable
7.
Mol Vis ; 7: 20-6, 2001 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-11182022

RESUMEN

PURPOSE: Corn1 is an autosomal recessive mutation characterized by corneal epithelial hyperplasia and stromal neovascularization. The aim of the present study is to examine the expression patterns of specific epithelial and stromal proteins in corn/corn1 mutant mice. METHODS: Immunohistochemistry with antibodies directed against keratins 1, 4, 5, 12, and 14 as well as loricrin, filaggrin, and involucrin were performed in corn1/corn1 and wild type, A.By/SnJ strain, mice at 4 weeks of age. Western blot hybridization was performed to confirm the presence of involucrin in corneas. In situ and northern blot hybridization were used to evaluate the expression of keratin 12, lumican, and keratocan in these mice. RESULTS: In corn1/corn1 mice, focal areas of corneal epithelial hyperplasia alternate with epithelium with normal appearance. Both regions of normal and hyperplastic corneal epithelium were labeled by anti-keratin 12 antibodies through all corneal epithelial layers. The anti-keratin 14 antibody only labeled the basal cell layer in normal epithelial areas, whereas it labeled both basal and suprabasal cell layers in hyperplastic areas. In wild type mice, anti-keratin 12 antibodies labeled all corneal epithelial layers, whereas anti-keratin 14 labeled the basal corneal epithelial cells only. Positive staining by anti-involucrin antibody was demonstrated in the basal corneal epithelial layer of wild type mice and normal areas of corn1/corn1 mice. Similarly, as observed with anti-keratin 14 antibody, the anti-involucrin antibody labeled both basal and suprabasal cell layers of hyperplastic corneal epithelium of corn1/corn1 mice. Antibodies against keratin 1, keratin 4, loricrin, and fillagrin did not label the corneas of wild type mice or corn1/corn1 mice. Northern hybridization indicated that the expressions of keratocan and lumican mRNA levels were up regulated in corn1/corn1 mice, but keratin 12 mRNA remained similar to that of the wild type mice. In situ hybridization revealed that the lumican mRNA was detected in epithelial and stromal cells of corn1/corn1 mice, whereas keratocan mRNA was only detected in stromal cells. CONCLUSIONS: Hyperproliferative epithelial cells of corn1/corn1 mice have increased levels of expression of keratin 14 and involucrin, but do not exhibit the phenotypical characteristics of cornification. These observations indicate that factors associated with the phenotypes of corn1/corn1 mice do not alter the cornea-type epithelial differentiation of keratin 12 expression, but cause aberrant expression of lumican by corneal epithelial cells.


Asunto(s)
Neovascularización de la Córnea/metabolismo , Sustancia Propia/metabolismo , Epitelio Corneal/metabolismo , Proteínas del Ojo/genética , Ratones Mutantes/metabolismo , Animales , Northern Blotting , Western Blotting , Proteoglicanos Tipo Condroitín Sulfato/genética , Proteoglicanos Tipo Condroitín Sulfato/metabolismo , Neovascularización de la Córnea/genética , Neovascularización de la Córnea/patología , Sustancia Propia/irrigación sanguínea , Epitelio Corneal/patología , Proteínas del Ojo/metabolismo , Fibroblastos/metabolismo , Proteínas Filagrina , Expresión Génica , Hiperplasia , Hibridación in Situ , Proteínas de Filamentos Intermediarios/genética , Proteínas de Filamentos Intermediarios/metabolismo , Sulfato de Queratano/genética , Sulfato de Queratano/metabolismo , Queratinas/genética , Queratinas/metabolismo , Lumican , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Mutantes/genética , Precursores de Proteínas , ARN Mensajero/biosíntesis
8.
Brain Res Mol Brain Res ; 81(1-2): 109-17, 2000 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-11000483

RESUMEN

The tubby gene family consists of four members, TUB, TULP1, TULP2 and TULP3, with unknown function. However, a splice junction mutation within the mouse tub gene leads to retinal and cochlear degeneration, as well as maturity onset obesity and insulin resistance. Mutations within human TULP1 have also been shown to co-segregate in several cases of autosomal recessive retinitis pigmentosa (RP) and TULP1 deficiency in mice leads to retinal degeneration. The primary amino acid sequences of the tubby family members do not predict a likely biochemical function. As a first step in defining their function, we present a detailed characterization of the cellular and subcellular localization of the human (TUB) and mouse (tub) homologous gene products. We report the isolation of TUB splice variants which have different subcellular localizations (nuclear versus cytoplasmic) and which define a nuclear localization signal. In addition, using green fluorescent protein (GFP) tags, we observe a nuclear localization for TULP1, similar to TUB splicing forms TUB 561 and TUB 506. Finally, we report tubby expression in mouse brain by in situ hybridization and by immunohistochemistry with polyclonal antibodies. Protein was found in both the hypothalamic satiety centers and in a variety of other CNS structures including the cortex, cerebellum, olfactory bulb and hippocampus. Both nuclear and cytoplasmic signals were detected with a series of independently generated polyclonal antibodies, consistent with the presence of multiple alternatively spliced isoforms within the CNS.


Asunto(s)
Encéfalo/metabolismo , Proteínas del Ojo/genética , Familia de Multigenes , Proteínas/genética , Testículo/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Secuencia de Aminoácidos , Animales , Células COS , Caenorhabditis elegans , Chlorocebus aethiops , Secuencia Conservada , Perros , Drosophila , Exones , Proteínas del Ojo/análisis , Genes Reporteros , Proteínas Fluorescentes Verdes , Humanos , Péptidos y Proteínas de Señalización Intercelular , Péptidos y Proteínas de Señalización Intracelular , Proteínas Luminiscentes/análisis , Proteínas Luminiscentes/genética , Masculino , Datos de Secuencia Molecular , Obesidad/genética , Proteínas/análisis , Proteínas Recombinantes de Fusión/análisis , Proteínas Recombinantes de Fusión/biosíntesis , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Transfección
9.
Invest Ophthalmol Vis Sci ; 41(10): 3149-57, 2000 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10967077

RESUMEN

PURPOSE: To characterize the genetics and phenotype of a new mouse mutant with retinal degeneration, rd6, that is associated with extensive, scattered, small white retinal dots seen ophthalmoscopically. METHODS: The phenotype was characterized using ophthalmoscopy, fundus photography, electroretinography, light microscopy, immunocytochemistry, and electron microscopy. Genetic characterization and linkage analysis studies were performed using standard methods. RESULTS: The inheritance pattern of rd6 is autosomal recessive. Linkage analysis mapped rd6 to mouse Chromosome 9 approximately 24 cM from the centromere, suggesting that the human homolog may be on chromosome 11q23. Ophthalmoscopic examination of mice homozygous for rd6 revealed discrete subretinal spots oriented in a regular pattern across the retina. The retinal spots appeared by 8 to 10 weeks of age and persisted through advanced stages of retinal degeneration. Histologic examination revealed large cells in the subretinal space, typically juxtaposed to the retinal pigment epithelium. The white dots seen on fundus examination corresponded both in distribution and size to these large cells. By 3 months of age, the cells were filled with membranous profiles, lipofuscin-like material, and pigment. These cells reacted strongly with an antibody directed against a mouse macrophage-associated antigen. Photoreceptor cells progressively degenerated with age, and an abnormal electroretinogram was initially detected between 1 and 2 months of age. CONCLUSIONS: The fundi of mice homozygous for rd6 exhibit phenotypic similarities to the human flecked retinal disorder retinitis punctata albescens. Thus, rd6/rd6 mice may be a model for understanding the etiology of this or similar disorders. The relationship between the aberrant subretinal cells and the concomitant photoreceptor degeneration remains to be established.


Asunto(s)
Modelos Animales de Enfermedad , Ceguera Nocturna/genética , Células Fotorreceptoras de Vertebrados/ultraestructura , Degeneración Retiniana/genética , Animales , Mapeo Cromosómico , Cromosomas/genética , Electrorretinografía , Femenino , Técnica del Anticuerpo Fluorescente Indirecta , Ligamiento Genético , Masculino , Ratones , Ratones Endogámicos C3H , Ceguera Nocturna/fisiopatología , Oftalmoscopía , Fenotipo , Células Fotorreceptoras de Vertebrados/fisiología , Degeneración Retiniana/patología , Degeneración Retiniana/fisiopatología
10.
Invest Ophthalmol Vis Sci ; 41(8): 2352-6, 2000 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10892883

RESUMEN

PURPOSE: To localize tubby-like protein 1 (TULP1) in developing and adult human retinas. METHODS: TULP1 was localized by immunofluorescence microscopy in human retinas, aged 8.4 fetal weeks to adult. TULP1-positive cells were identified by double labeling with antibodies specific for cones, rods, and astrocytes. RESULTS: In adult retinas, anti-TULP1 labels cone and rod inner segments, somata, and synapses; outer segments are TULP1-negative. A few inner nuclear and ganglion cells are weakly TULP1-positive. In fetal retinas, cells at the outer retinal border are TULP1-positive at 8.4 weeks. At 11 weeks, the differentiating central cones are strongly TULP1-reactive and some are positive for blue cone opsin. At 15.4 weeks, all central cones are strongly positive for TULP1 and many are reactive for red/green cone opsin. At 17.4 weeks, central rods are weakly TULP-reactive. In peripheral retina at 15.4 weeks to 1 month after birth, displaced cones in the nerve fiber layer are positive for TULP1, recoverin, and blue cone opsin. Some ganglion cells are weakly reactive for TULP1 at 11 weeks and later, but astrocytes and the optic nerve are TULP1-negative at all ages examined. CONCLUSIONS: The finding of TULP1 labeling of cones before they are reactive for blue or red/green cone opsin suggests an important role for TULP1 in development. TULP1 expression in both developing and mature cones and rods is consistent with a primary photoreceptor defect in retinitis pigmentosa (RP) caused by TULP1 mutations. Weak TULP1-immunolabeling of some inner retinal neurons in developing and adult retinas suggests that optic disc changes in patients with RP who have TULP1 mutations may be primary as well as secondary to photoreceptor degeneration.


Asunto(s)
Proteínas del Ojo/análisis , Retina/química , Retina/crecimiento & desarrollo , Adulto , Astrocitos/química , Feto/química , Técnica del Anticuerpo Fluorescente Indirecta , Humanos , Lactante , Recién Nacido , Microscopía Fluorescente , Células Fotorreceptoras de Vertebrados/química
11.
Proc Natl Acad Sci U S A ; 97(10): 5551-6, 2000 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-10805811

RESUMEN

The rd7 mouse, an animal model for hereditary retinal degeneration, has some characteristics similar to human flecked retinal disorders. Here we report the identification of a deletion in a photoreceptor-specific nuclear receptor (mPNR) mRNA that is responsible for hereditary retinal dysplasia and degeneration in the rd7 mouse. mPNR was isolated from a pool of photoreceptor-specific cDNAs originally created by subtractive hybridization of mRNAs from normal and photoreceptorless rd mouse retinas. Localization of the gene corresponding to mPNR to mouse Chr 9 near the rd7 locus made it a candidate for the site of the rd7 mutation. Northern analysis of total RNA isolated from rd7 mouse retinas revealed no detectable signal after hybridization with the mPNR cDNA probe. However, with reverse transcription-PCR, we were able to amplify different fragments of mPNR from rd7 retinal RNA and to sequence them directly. We found a 380-nt deletion in the coding region of the rd7 mPNR message that creates a frame shift and produces a premature stop codon. This deletion accounts for more than 32% of the normal protein and eliminates a portion of the DNA-binding domain. In addition, it may result in the rapid degradation of the rd7 mPNR message by the nonsense-mediated decay pathway, preventing the synthesis of the corresponding protein. Our findings demonstrate that mPNR expression is critical for the normal development and function of the photoreceptor cells.


Asunto(s)
Mapeo Cromosómico , Células Fotorreceptoras de Vertebrados/fisiología , ARN Mensajero/genética , Receptores Citoplasmáticos y Nucleares/genética , Degeneración Retiniana/genética , Factores de Transcripción , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Codón de Terminación , Cartilla de ADN , Electrorretinografía , Marcadores Genéticos , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Ratones Mutantes , Datos de Secuencia Molecular , Receptores Nucleares Huérfanos , Células Fotorreceptoras de Vertebrados/patología , Degeneración Retiniana/patología , Degeneración Retiniana/fisiopatología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Eliminación de Secuencia
12.
Hum Mol Genet ; 9(2): 155-63, 2000 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-10607826

RESUMEN

The tub gene is a member of a small, well conserved neuronal gene family of unknown function. Mutations within this gene lead to early-onset blindness and deafness, as well as late-onset obesity and insulin resistance. To test the hypothesis that mutations within other members of this gene family would lead to similar phenotypes as observed in tubby mice, and hence have similar functional properties, we have generated null mutants of the tubby-like protein ( Tulp ) 1 gene by homologous recombination. Similarly to tubby mice, Tulp1 (-/-)mice exhibit an early-onset retinal degeneration with a progressive, rapid loss of photoreceptors, further supporting the notion that previously identified mutations within the human TULP1 gene are indeed causative of retinitis pigmentosa. However, in contrast to tubby mice, Tulp1 (-/-)mice exhibited normal hearing ability and, surprisingly, normal body weight despite the fact that both TUB and TULP1 are expressed in the same neurons within the hypothalamus in areas known to be involved in feeding behavior and energy homeo stasis. However, TUB and TULP1 show a distinctly different staining pattern in the nucleus of these neurons, perhaps explaining the difference in body weight between the Tulp1 (-/-)and tubby mutant mice.


Asunto(s)
Proteínas del Ojo/genética , Mutación/genética , Obesidad/genética , Degeneración Retiniana/genética , Animales , Química Encefálica/genética , Proteínas del Ojo/biosíntesis , Fondo de Ojo , Células Ciliadas Auditivas Internas/patología , Pruebas Auditivas , Humanos , Hipotálamo/metabolismo , Etiquetado Corte-Fin in Situ , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oftalmoscopía , Retina/patología , Retina/ultraestructura , Degeneración Retiniana/patología , Aumento de Peso/genética
13.
Hum Genet ; 105(5): 474-9, 1999 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-10598815

RESUMEN

Alström syndrome is a rare autosomal recessive disorder characterized by retinal degeneration, sensorineural hearing loss, early-onset obesity, and non-insulin-dependent diabetes mellitus. The gene for Alström syndrome (ALMS1) has been previously localized to human chromosome 2p13 by homozygosity mapping in two distinct isolated populations - French Acadian and North African. Pair-wise analyses resulted in maximum lod (logarithm of the odds ratio) scores of 3.84 and 2.9, respectively. To confirm these findings, a large linkage study was performed in twelve additional families segregating for Alström syndrome. A maximum two-point lod score of 7.13 (theta = 0.00) for marker D2S2110 and a maximum cumulative multipoint lod score of 9.16 for marker D2S2110 were observed, further supporting linkage to chromosome 2p13. No evidence of genetic heterogeneity was observed in these families. Meiotic recombination events have localized the critical region containing ALMS1 to a 6.1-cM interval flanked by markers D2S327 and D2S286. A fine resolution radiation hybrid map of 31 genes and markers has been constructed.


Asunto(s)
Cromosomas Humanos Par 2/genética , Diabetes Mellitus Tipo 2/genética , Ligamiento Genético , Pérdida Auditiva Sensorineural/genética , Obesidad/genética , Degeneración Retiniana/genética , Secuencia de Bases , Mapeo Cromosómico , Cartilla de ADN/genética , Femenino , Genes Recesivos , Marcadores Genéticos , Humanos , Escala de Lod , Masculino , Linaje , Síndrome
14.
Invest Ophthalmol Vis Sci ; 40(11): 2706-12, 1999 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-10509669

RESUMEN

PURPOSE: The family of tubby-like proteins (TULPs), consisting of four family members, are all expressed in-the retina at varying levels. Mutations within two members, tub and TULP1, are known to lead to retinal degeneration in mouse and humans, respectively, suggesting the functional importance of this family of proteins in the retina. Despite a high degree of conservation in the carboxy-terminal region (e.g., putative functional domain of the genes) among family members, they are unable to compensate for one another. The purpose of this study was to provide a rationale for this lack of compensation by investigating the spatial distribution of tubby gene family members in the retina and to investigate the mechanism of photoreceptor cell death in tubby mice. METHODS: In situ hybridization using riboprobes specific for each tubby gene family member and immunohistochemistry for TUB and TULP1 were performed to determine their expression patterns in the retina of tubby and wild-type control mice. The terminal dUTP nick-end labeling (TUNEL) assay was performed to detect apoptotic cells in the retina of tubby and wild-type control mice. RESULTS: tub mRNA was found to be expressed throughout the retina, with highest expression in the ganglion cell layer (GCL) and photoreceptor cells. In contrast, Tulp1 expression was observed only in photoreceptor cells and Tulp3 mRNA was expressed at a moderate level only in the inner nuclear layer (INL) and GCL. The results of the immunohistochemical analysis paralleled those observed in the in situ studies. TUB immunoreactivity was most highly concentrated in the GCL, in the inner and outermost regions of the INL, in the outer plexiform layer (OPL), and in the inner segments of photoreceptor cells. Similarly, TULP1 immunoreactivity was observed in the OPL and inner segments of the photoreceptor cells. No differences in expression at the mRNA or protein level were observed for any of the molecules tested in tubby or wild-type mice. TUNEL-positive cells were detected in the ONL of tubby mice, whereas very few were seen in the same layer of age-matched control mice. CONCLUSIONS: Although all tubby gene family members are expressed in the retina, they each have different cell-specific expression patterns, which may account in part for their inability to compensate for the loss of one family member. The photoreceptor cell death in tubby mice occurs through an apoptotic mechanism, which is known to be the common final outcome of other forms of retinal degeneration.


Asunto(s)
Proteínas del Ojo/genética , Expresión Génica , Proteínas/genética , Retina/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Apoptosis/fisiología , Proteínas del Ojo/metabolismo , Técnicas para Inmunoenzimas , Hibridación in Situ , Etiquetado Corte-Fin in Situ , Péptidos y Proteínas de Señalización Intercelular , Péptidos y Proteínas de Señalización Intracelular , Ratones , Ratones Mutantes , Células Fotorreceptoras de Vertebrados/citología , Células Fotorreceptoras de Vertebrados/fisiología , Proteínas/metabolismo , ARN Mensajero/metabolismo , Retina/citología
15.
Hum Mol Genet ; 8(9): 1761-7, 1999 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10441341

RESUMEN

Quantitative trait locus (QTL) analysis of genetic crosses has proven to be a useful tool for identifying loci associated with specific phenotypes and for dissecting genetic components of complex traits. Inclusion of a mutation that interacts epistatically with QTLs in genetic crosses is a unique and potentially powerful method of revealing the function of novel genes and pathways. Although we know that a mutation within the novel tub gene leads to obesity and cochlear and retinal degeneration, the biological function of the gene and the mechanism by which it induces its phenotypes are not known. In the current study, a QTL analysis for auditory brainstem response (ABR) thresholds, which indicates hearing ability, was performed in tubby mice from F(2)intercrosses between C57BL/6J- tub / tub and AKR/J-+/+ F(1)hybrids (AKR intercross) and between C57BL/6J- tub / tub and CAST/Ei.B6- tub / tub F(1)hybrids (CAST intercross). A major QTL, designated asmodifieroftubbyhearing1 ( moth1 ), was identified on chromosome 2 with a LOD score of 33.4 ( P < 10(-33)) in the AKR intercross (181 mice) and of 6.0 ( P < 10(-6)) in the CAST intercross (46 mice). This QTL is responsible for 57 and 43% of ABR threshold variance, respectively, in each strain combination. In addition, a C57BL/6J congenic line carrying a 129/Ola segment encompassing the described QTL region when made homozygous for tubby also exhibits normal hearing ability. We hypothesize that C57BL/6J carries a recessive mutation of the moth1 gene which interacts with the tub mutation to cause hearing loss in tub / tub mice. A moth1 allele from either AKR/J, CAST/Ei or 129/Ola is sufficient to protect C57BL/6J- tub / tub mice from hearing loss.


Asunto(s)
Trastornos de la Audición/genética , Proteínas/genética , Proteínas Adaptadoras Transductoras de Señales , Animales , Mapeo Cromosómico , Cóclea/citología , Cruzamientos Genéticos , Potenciales Evocados Auditivos del Tronco Encefálico/genética , Técnica del Anticuerpo Fluorescente , Ligamiento Genético , Genotipo , Escala de Lod , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Carácter Cuantitativo Heredable
16.
J Lipid Res ; 40(7): 1328-35, 1999 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10393218

RESUMEN

Mice from the inbred strain C57BLKS/J (BKS) exhibit increased susceptibility to both diabetes and atherosclerosis compared to C57BL/6J (B6) mice. To determine whether the differences in diabetes and atherosclerosis are related, we carried out a cross between B6-db/db and BKS. We selected 99 female F2-db/db progeny, tested the progeny for plasma lipids, plasma glucose, and fatty-streak lesions, and used quantitative trait loci (QTL) analysis to identify the chromosomal regions associated with these phenotypes. No major QTL were found for total cholesterol, VLDL-cholesterol, or triglycerides. Two suggestive QTL were found for HDL-cholesterol (LOD scores of 2. 7 and 2.8), and two suggestive loci were found for plasma glucose (LOD scores of 2.3 and 2.0). Lesion size was not correlated with plasma lipid levels or glucose. Lesion size was determined by a locus at D12Mit49 with a LOD score of 2.5 and a significant likelihood ratio statistic. The gene for apolipoprotein apoB lies within the region, but apoB levels were similar in strains B6 and BKS. The QTL on Chr 12 was confirmed by constructing a congenic strain with BKS alleles in the QTL region on a B6 genetic background. We conclude that susceptibilities to diabetes and atherosclerosis are not conferred by the same genes in these strains and that a major gene on Chr 12, which we name Ath6, determines the difference in atherosclerosis susceptibility.


Asunto(s)
Arteriosclerosis/genética , Diabetes Mellitus/genética , Predisposición Genética a la Enfermedad/genética , Ratones Endogámicos C57BL/genética , Carácter Cuantitativo Heredable , Alelos , Animales , Aorta/patología , Apolipoproteínas B/genética , Arteriosclerosis/patología , Glucemia/análisis , Mapeo Cromosómico , Dieta Aterogénica , Femenino , Lípidos/sangre , Masculino , Ratones
17.
Invest Ophthalmol Vis Sci ; 40(8): 1874-8, 1999 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10393064

RESUMEN

PURPOSE: To demonstrate the importance of genetic background interaction on the development of ocular phenotypes in p53-deficient mice. METHODS: Eyes of adult mice, homozygous and heterozygous for the p53 gene disruption in the 129/SvJ and C57BL/6J (B6) genetic backgrounds, and their F1 progeny were examined by indirect ophthalmoscopy and by light microscopy. RESULTS: Indirect ophthalmoscopy revealed unilateral or bilateral vitreal opacities, fibrous retrolental tissue, and retinal folds in adult B6 mice but not in 129/Sv mice homozygous for a p53 null mutation. In B6 p53-/- mice, blood vessels extended from the peripapillary inner retina through the posterior vitreous and into the retrolental membrane. Optic nerves were hypoplastic. CONCLUSIONS: These findings indicate that alleles from the B6 background contribute to the aberrant ocular phenotypes observed in p53 deficiency. They also suggest that p53 or the pathway in which it functions may be important for normal eye development.


Asunto(s)
Anomalías Múltiples/genética , Anomalías del Ojo/genética , Genes p53/genética , Retina/anomalías , Proteína p53 Supresora de Tumor/deficiencia , Cuerpo Vítreo/anomalías , Anomalías Múltiples/patología , Animales , Catarata/genética , Catarata/patología , Cartilla de ADN/química , Anomalías del Ojo/patología , Oftalmopatías/genética , Oftalmopatías/patología , Femenino , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Nervio Óptico/anomalías , Nervio Óptico/patología , Fenotipo , Retina/patología , Displasia Retiniana/genética , Displasia Retiniana/patología , Proteína p53 Supresora de Tumor/genética , Cuerpo Vítreo/patología
18.
Arterioscler Thromb Vasc Biol ; 19(6): 1512-7, 1999 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10364083

RESUMEN

The relationship between atheroma lipid composition and serum lipoprotein and oxidation measurements has not been fully explored. To address this question, we studied serum, plasma, and aortic wall specimens from 66 subjects undergoing coronary artery bypass graft surgery. The lipid composition of aortic specimens was characterized in terms of cholesterol ester and cholesterol crystal plus phospholipid by using hot-stage polarizing light microscopy; tissue oxidation status was assessed by measuring conjugated dienes. Serum lipoprotein-related measurements included total cholesterol, triglyceride, low density lipoprotein (LDL) cholesterol, high density lipoprotein (HDL) cholesterol, apolipoproteins B and AI, and lipoprotein(a). Oxidation status was assessed by measuring LDL mobility, thiobarbituric acid-reactive substances, LDL conjugated dienes, and IgG and IgM autoantibodies against oxidized LDL. Fasting blood glucose was also determined. Lesion cholesterol crystal plus phospholipid content was associated inversely with serum HDL cholesterol levels (r=-0.279, P=0.029) and positively with fasting blood glucose (r=0.359, P=0.016), LDL mobility (0.276, P<0.05), and IgM autoantibodies against oxidized LDL (r=0.272, P=0.037). There was also a significant relationship between the level of aortic tissue conjugated dienes and plasma LDL mobility (r=0.332, P=0.007). In multivariate analysis, IgM autoantibodies against oxidized LDL, fasting blood glucose, and LDL mobility, in descending order of significance, together accounted for 35% of the variability in aortic lesion cholesterol crystal plus phospholipid content. These data support direct and independent roles for oxidation and hyperglycemia in the pathophysiology of atherosclerosis.


Asunto(s)
Arteriosclerosis/metabolismo , Puente de Arteria Coronaria , Lípidos/análisis , Lipoproteínas/metabolismo , Anciano , Autoanticuerpos/sangre , Ésteres del Colesterol/metabolismo , Femenino , Humanos , Inmunoglobulina M/sangre , Lipoproteínas LDL/inmunología , Masculino , Persona de Mediana Edad , Oxidación-Reducción
20.
Genomics ; 53(3): 359-64, 1998 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-9799602

RESUMEN

The human dynactin 1 gene (DCTN1) is positioned on chromosome 2p13, the candidate region for various diseases including Alström syndrome, limb-girdle muscle dystrophy, and Miyoshi myopathy. Here, we report the exon-intron structure of DCTN1 along with characterization of the 5' upstream sequence and alternative splice variants previously identified by Tokito et al. (1996), Mol. Biol. Cell 7: 1167-1180). Knowledge of the genomic structure of DCTN1allowed us to design intronic primers necessary for analyzing mutations in families segregating for diseases linked to this gene. These primers were tested on a French Acadian kindred segregating for Alström syndrome. No mutations were observed within the coding region of DCTN1 in this family. However, the intronic primers should allow for the rapid amplification of the coding region for mutational analysis of additional Alström families and other diseases tightly linked to the DCTN1locus on chromosome 2p13.


Asunto(s)
Pérdida Auditiva Sensorineural/genética , Proteínas Asociadas a Microtúbulos/genética , Obesidad/genética , Degeneración Retiniana/genética , Empalme Alternativo , Secuencia de Aminoácidos , Secuencia de Bases , Niño , Cromosomas Humanos Par 2/genética , Análisis Mutacional de ADN , Cartilla de ADN/genética , Complejo Dinactina , Exones , Femenino , Variación Genética , Humanos , Intrones , Masculino , Datos de Secuencia Molecular , Síndrome
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...